Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Identification of a motor-to-auditory pathway important for vocal learning

Abstract

Learning to vocalize depends on the ability to adaptively modify the temporal and spectral features of vocal elements. Neurons that convey motor-related signals to the auditory system are theorized to facilitate vocal learning, but the identity and function of such neurons remain unknown. Here we identify a previously unknown neuron type in the songbird brain that transmits vocal motor signals to the auditory cortex. Genetically ablating these neurons in juveniles disrupted their ability to imitate features of an adult tutor's song. Ablating these neurons in adults had little effect on previously learned songs but interfered with their ability to adaptively modify the duration of vocal elements and largely prevented the degradation of songs' temporal features that is normally caused by deafening. These findings identify a motor to auditory circuit essential to vocal imitation and to the adaptive modification of vocal timing.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: A distinct class of projection neurons links the song premotor nucleus HVC to the auditory region Av.
Figure 2: HVCAv neurons transmit motor-related signals during song production.
Figure 3: HVCAv neurons receive selective input from premotor HVCRA neurons.
Figure 4: Intersectional genetic ablation of HVCAv neurons in juvenile zebra finches impairs their ability to copy a tutor song.
Figure 5: Intersectional ablation of HVCAv neurons in adult birds does not disrupt song production but does attenuate deafening-induced degradation of song's temporal features.
Figure 6: Intersectional ablation of HVCAv neurons in adult zebra finches interferes with feedback-dependent plasticity of song element timing but not syllable pitch.

Similar content being viewed by others

References

  1. Eliades, S.J. & Wang, X. Neural substrates of vocalization feedback monitoring in primate auditory cortex. Nature 453, 1102–1106 (2008).

    Article  CAS  PubMed  Google Scholar 

  2. Schneider, D.M., Nelson, A. & Mooney, R. A synaptic and circuit basis for corollary discharge in the auditory cortex. Nature 513, 189–194 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Lee, S., Kruglikov, I., Huang, Z.J., Fishell, G. & Rudy, B. A disinhibitory circuit mediates motor integration in the somatosensory cortex. Nat. Neurosci. 16, 1662–1670 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Niell, C.M. & Stryker, M.P. Modulation of visual responses by behavioral state in mouse visual cortex. Neuron 65, 472–479 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Wang, J. et al. Action planning and predictive coding when speaking. Neuroimage 91, 91–98 (2014).

    Article  PubMed  Google Scholar 

  6. Hickok, G., Houde, J. & Rong, F. Sensorimotor integration in speech processing: computational basis and neural organization. Neuron 69, 407–422 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Crapse, T.B. & Sommer, M.A. Corollary discharge across the animal kingdom. Nat. Rev. Neurosci. 9, 587–600 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Houde, J.F. & Chang, E.F. The cortical computations underlying feedback control in vocal production. Curr. Opin. Neurobiol. 33, 174–181 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Cullen, K.E. Sensory signals during active versus passive movement. Curr. Opin. Neurobiol. 14, 698–706 (2004).

    Article  CAS  PubMed  Google Scholar 

  10. Sperry, R.W. Neural basis of the spontaneous optokinetic response produced by visual inversion. J. Comp. Physiol. Psychol. 43, 482–489 (1950).

    Article  CAS  PubMed  Google Scholar 

  11. Poulet, J.F. & Hedwig, B. The cellular basis of a corollary discharge. Science 311, 518–522 (2006).

    Article  CAS  PubMed  Google Scholar 

  12. von Holst, E. & Mittelstaedt, H. Das reaferenzprinzip. [The principle of reafference]. Naturwissenschaften 37, 464–476 (1950).

    Article  Google Scholar 

  13. Bell, C.C. An efference copy which is modified by reafferent input. Science 214, 450–453 (1981).

    Article  CAS  PubMed  Google Scholar 

  14. Doupe, A.J. & Kuhl, P.K. Birdsong and human speech: common themes and mechanisms. Annu. Rev. Neurosci. 22, 567–631 (1999).

    Article  CAS  PubMed  Google Scholar 

  15. Immelmann, K. in Bird Vocalisations (ed. Hinde, R.A.) 61–74 (Cambridge University Press, 1969).

  16. Ali, F. et al. The basal ganglia is necessary for learning spectral, but not temporal, features of birdsong. Neuron 80, 494–506 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Eales, L.A. Song learning in zebra finches: some effects of song model availability on what is learnt and when. Anim. Behav. 33, 1293–1300 (1985).

    Article  Google Scholar 

  18. Tumer, E.C. & Brainard, M.S. Performance variability enables adaptive plasticity of 'crystallized' adult birdsong. Nature 450, 1240–1244 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Roberts, T.F., Gobes, S.M., Murugan, M., Ölveczky, B.P. & Mooney, R. Motor circuits are required to encode a sensory model for imitative learning. Nat. Neurosci. 15, 1454–1459 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Long, M.A. & Fee, M.S. Using temperature to analyse temporal dynamics in the songbird motor pathway. Nature 456, 189–194 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Vallentin, D., Kosche, G., Lipkind, D. & Long, M.A. Neural circuits. Inhibition protects acquired song segments during vocal learning in zebra finches. Science 351, 267–271 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Keller, G.B. & Hahnloser, R.H. Neural processing of auditory feedback during vocal practice in a songbird. Nature 457, 187–190 (2009).

    Article  CAS  PubMed  Google Scholar 

  23. Heinks-Maldonado, T.H., Mathalon, D.H., Gray, M. & Ford, J.M. Fine-tuning of auditory cortex during speech production. Psychophysiology 42, 180–190 (2005).

    Article  PubMed  Google Scholar 

  24. Hahnloser, R.H., Kozhevnikov, A.A. & Fee, M.S. An ultra-sparse code underlies the generation of neural sequences in a songbird. Nature 419, 65–70 (2002).

    Article  CAS  PubMed  Google Scholar 

  25. Hamaguchi, K., Tanaka, M. & Mooney, R. A distributed recurrent network contributes to temporally precise vocalizations. Neuron 91, 680–693 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Alvarez-Buylla, A., Kirn, J.R. & Nottebohm, F. Birth of projection neurons in adult avian brain may be related to perceptual or motor learning. Science 249, 1444–1446 (1990).

    Article  CAS  PubMed  Google Scholar 

  27. Sohrabji, F., Nordeen, E.J. & Nordeen, K.W. Selective impairment of song learning following lesions of a forebrain nucleus in the juvenile zebra finch. Behav. Neural Biol. 53, 51–63 (1990).

    Article  CAS  PubMed  Google Scholar 

  28. Scharff, C. & Nottebohm, F. A comparative study of the behavioral deficits following lesions of various parts of the zebra finch song system: implications for vocal learning. J. Neurosci. 11, 2896–2913 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Akutagawa, E. & Konishi, M. New brain pathways found in the vocal control system of a songbird. J. Comp. Neurol. 518, 3086–3100 (2010).

    Article  PubMed  Google Scholar 

  30. Mooney, R. Different subthreshold mechanisms underlie song selectivity in identified HVc neurons of the zebra finch. J. Neurosci. 20, 5420–5436 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tschida, K.A. & Mooney, R. Deafening drives cell-type-specific changes to dendritic spines in a sensorimotor nucleus important to learned vocalizations. Neuron 73, 1028–1039 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wild, J.M., Williams, M.N., Howie, G.J. & Mooney, R. Calcium-binding proteins define interneurons in HVC of the zebra finch (Taeniopygia guttata). J. Comp. Neurol. 483, 76–90 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. Dutar, P., Vu, H.M. & Perkel, D.J. Multiple cell types distinguished by physiological, pharmacological, and anatomic properties in nucleus HVc of the adult zebra finch. J. Neurophysiol. 80, 1828–1838 (1998).

    Article  CAS  PubMed  Google Scholar 

  34. Hattox, A.M. & Nelson, S.B. Layer V neurons in mouse cortex projecting to different targets have distinct physiological properties. J. Neurophysiol. 98, 3330–3340 (2007).

    Article  PubMed  Google Scholar 

  35. Kim, E.J., Juavinett, A.L., Kyubwa, E.M., Jacobs, M.W. & Callaway, E.M. Three types of cortical layer 5 neurons that differ in brain-wide connectivity and function. Neuron 88, 1253–1267 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Long, M.A., Jin, D.Z. & Fee, M.S. Support for a synaptic chain model of neuronal sequence generation. Nature 468, 394–399 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liberti, W.A., III et al. Unstable neurons underlie a stable learned behavior. Nat. Neurosci. 19, 1665–1671 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Picardo, M.A. et al. Population-level representation of a temporal sequence underlying song production in the zebra finch. Neuron 90, 866–876 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hamaguchi, K., Tschida, K.A., Yoon, I., Donald, B.R. & Mooney, R. Auditory synapses to song premotor neurons are gated off during vocalization in zebra finches. Elife 3, e01833 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Zhou, P., Resendez, S.L., Stuber, G.D., Kass, R.E. & Paninski, L. Efficient and accurate extraction of in vivo calcium signals from microendoscope video data. Preprint at https://arxiv.org/abs/1605.07266 (2016).

  41. Prather, J.F., Peters, S., Nowicki, S. & Mooney, R. Precise auditory-vocal mirroring in neurons for learned vocal communication. Nature 451, 305–310 (2008).

    Article  CAS  PubMed  Google Scholar 

  42. Mooney, R. & Prather, J.F. The HVC microcircuit: the synaptic basis for interactions between song motor and vocal plasticity pathways. J. Neurosci. 25, 1952–1964 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Yang, C.F. et al. Sexually dimorphic neurons in the ventromedial hypothalamus govern mating in both sexes and aggression in males. Cell 153, 896–909 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Nordeen, K.W. & Nordeen, E.J. Auditory feedback is necessary for the maintenance of stereotyped song in adult zebra finches. Behav. Neural Biol. 57, 58–66 (1992).

    Article  CAS  PubMed  Google Scholar 

  45. Brainard, M.S. & Doupe, A.J. Interruption of a basal ganglia-forebrain circuit prevents plasticity of learned vocalizations. Nature 404, 762–766 (2000).

    Article  CAS  PubMed  Google Scholar 

  46. Williams, H., Crane, L.A., Hale, T.K., Esposito, M.A. & Nottebohm, F. Right-side dominance for song control in the zebra finch. J. Neurobiol. 23, 1006–1020 (1992).

    Article  CAS  PubMed  Google Scholar 

  47. Bauer, E.E. et al. A synaptic basis for auditory-vocal integration in the songbird. J. Neurosci. 28, 1509–1522 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Nelson, A. et al. A circuit for motor cortical modulation of auditory cortical activity. J. Neurosci. 33, 14342–14353 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Mandelblat-Cerf, Y., Las, L., Denisenko, N. & Fee, M.S. A role for descending auditory cortical projections in songbird vocal learning. Elife 3 (2014).

  50. Gale, S.D., Person, A.L. & Perkel, D.J. A novel basal ganglia pathway forms a loop linking a vocal learning circuit with its dopaminergic input. J. Comp. Neurol. 508, 824–839 (2008).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank D. Schneider, K. Tschida, T. Warren and S. Lisberger for reading and commenting on the manuscript; K. Hamaguchi (Duke University Medical Center) for software support; W.Y. Peh (Duke University Medical Center) for help with calcium imaging experiments; B.P. Olveczky (Harvard University) for providing Conditional Auditory Feedback software; J. Baltzegar, M. Booze (Duke University Medical Center), J. Holdway and A. Guerrero (UT Southwestern Medical Center) for animal husbandry and laboratory support. This research was supported by grants from the National Science Foundation (R.M. (IOS-1354962), T.F.R. (IOS-1457206, IOS-1451034)), the US National Institutes of Health (R.M. (R01DC002524, R01NS099288), T.F.R. (R01DC014364), N.M.S. (R01NS049488, R01NS083872), M.J.K. (F30NS096871)), an Inscopix DECODE Award (R.M.), the Klingenstein-Simons Fellowship (T.F.R.), the Ellison Medical Foundation (N.M.S.), a JSPS Postdoctoral Fellowship for Research Abroad (M.T.), an Alpha Omega Alpha Research Fellowship (G.C.) and a NARSAD Young Investigator Grant (Essel Investigator) from the Brain & Behavior Research Foundation (T.F.R.).

Author information

Authors and Affiliations

Authors

Contributions

T.F.R., E.H. and R.M. conceived and designed all experiments. T.F.R. did the anatomical tracing experiments and juvenile genetic lesion experiments, and designed and oversaw the conditional auditory feedback experiments. E.H. did the anatomical tracing experiments, adult genetic lesioning experiments and adult deafening experiments. M.T., M.G.K. and G.C. did the slice physiology, calcium imaging and conditional auditory feedback experiments, respectively. C.F.Y. and N.M.S. provided virus used for cell ablation experiments. T.F.R. and R.M. wrote the manuscript. All authors read and commented on the manuscript.

Corresponding authors

Correspondence to Todd F Roberts or Richard Mooney.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Comparisons of GFP-expressing HVCAv, HVCRA and HVCX cell bodies.

a) Soma diameter measured at the long axis of the cell body for HVCAv (green), HVCRA (red), and HVCX (blue) cells (two-tailed t-test: HVCAv versus HVCRA, n= 29 for both groups, p = 0.0001; HVCAv versus HVCX, n = 29 for both groups, p = 0.0006). b) Soma area for HVCAv (green), HVCRA (red), and HVCX (blue) cells (two-tailed t-test: HVCAv versus HVCRA, n= 29 for both groups, p = 0.229; HVCAv versus HVCX, n = 29 for both groups, p < 0.0001). Numbers listed on top left corner of each panel indicate the mean and S.E.M. for each cell type. Inset shows Gaussian fits to data shown in histogram. HVCRA and HVCX cell body measurements derived from data published in Tschida and Mooney, 2012.

Supplementary Figure 2 Different HVCAv neurons show different activity patterns during singing in hearing-intact and deafened birds.

a) Field of GCaMP-labelled HVCAv neurons imaged through miniature microscope in a singing zebra finch in a single bird with hearing intact (left) and after deafening (right). Individual cells are indicated by colored outlines. Scale bars, 100 μm x 100 μm. b) Mean singing-related activity traces of five putative HVCAv neurons identified by CNMF analysis with hearing intact (left, n = 24 songs) and after deafening (right, n = 25 songs). Black lines represent mean activity traces for each neuron and the shaded area denotes ± one standard error of the mean response. Vertical scale bars, 10 (left) and 5 (right) arbitrary activity units; horizontal scale bars = 1 s and apply to traces in b – d; song motif onset is marked by vertical grey line. c) The summed activity from individual putative cell bodies shown in panel b). Vertical scale bars: 25 (left) and 20 (right) arbitrary activity units. d) Mean change in bulk fluorescence signals measured during singing from all birds with hearing intact (left, n = 4 birds) and after deafening (right, n = 2 birds) (same data as plotted in Figure 2c; scale bar 2% df/f). e) Cumulative probability distributions of mean decay time constants measured in singing zebra finches for GCaMP6s-expressing HVCAv neurons (blue, n = 16 ROIs) and a mixed population of GCaMP6s-expressing HVC neurons (red, n = 43 ROIs). The KS test p value is 0.9914 and the KS statistic is 0.1221. Raw imaging files to which the CNMF algorithm was applied are provided as Supplementary Movies 1 and 2.

Supplementary Figure 3 Effects of electrical stimulation on different HVC cell types.

a) Evoked EPSC in HVCAv cell after HVCRA stimulation at 80 (black trace) and 160 (grey trace) μA; scale bar 20 ms, 200 pA. b) Evoked IPSC in HVCAv cell after HVCRA stimulation at 80 uA; scale bar 20 ms, 300 pA. c) Evoked IPSC in HVCRA cell after HVCX stimulation at 80 μA; scale bar 20 ms, 50 pA. d) Evoked IPSC in HVCX cell after HVCX stimulation at 80 μA; scale bar 20 ms, 100 pA. e) Recording in HVCAv cell after electrical stimulation of LaM at 160 μA; scale bar 20 ms, 100 pA.

Supplementary Figure 4 Spectral derivatives illustrating learning outcome for all 5 (a–e) HVCAv lesioned birds.

Birds in panels a-c were tutored by the same adult male zebra finch in separate experiments. In panels a-d examples of copied syllable(s) are marked by a red line. The fifth zebra finch, in panel e, did not copy either of the two complex syllables in the tutors’ song. Birds and in panels a-b are the same birds illustrated in Figure 3 of the manuscript. Scale bar = 100ms (illustrated in bottom right of panel a).

Supplementary Figure 5 Cre-dependent Caspase 3 is efficacious and specific to targeted population.

a) Example of song from one bird before and after unilateral lesion of HVC from co-injection of Cre-dependent caspase 3 and Cre into HVC. b) Percent similarity to bird’s own song (self-similarity) before and after unilateral lesions of HVC for 2 birds after co-injection of Cre-dependent caspase 3 and Cre into HVC unilaterally. c) Average number of cells per 50 micron section of HVC in 2 birds in which HVCAv cells were unilaterally lesioned. HVCX cells, which were not targeted with the intersectional ablation strategy (blue), were comparable in number between unlesioned and lesioned hemispheres while HVCAv cells, which were targeted (red), showed a reduction by nearly half in lesioned hemispheres.

Supplementary Figure 6 Ablation of HVCAv in adult birds causes slight decreases in motif similarity and duration.

a) Experimental birds show a significant but slight decrease in percent similarity to bird’s own song (self-similarity) after HVCAv ablation (n = 12 HVCAv lesioned birds, paired two-sample t(11)=4.34, P = 0.001 comparing percent self-similarity before lesioning to percent self-similarity after lesioning in experimental birds.) b) Experimental birds show a significant decrease in motif duration after HVCAv ablation (n = 12 HVCAv lesioned birds, n = 12 HVCAv intact birds; two-sample t(22) = 2.49, P = 0.02 comparing percent change in motif duration from lesioned versus intact birds). c) Example sonograms of single syllables from two control birds before (top row) and after (bottom row) deafening. Scale bars, 50 ms. d) Example sonograms of single syllables from four experimental birds before (top row) and ten weeks after (bottom row) deafening. Scale bars, 50 ms.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Roberts, T., Hisey, E., Tanaka, M. et al. Identification of a motor-to-auditory pathway important for vocal learning. Nat Neurosci 20, 978–986 (2017). https://doi.org/10.1038/nn.4563

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4563

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing